1
|
Xu F, Sang J, Wang N, Wang M, Huang Y, Ma J, Chen H, Xie Q, Wei Z, Ye X. Microwave ablation combined with immune checkpoint inhibitor enhanced the antitumor immune activation and memory in rechallenged tumor mouse model. Cancer Immunol Immunother 2025; 74:161. [PMID: 40131498 PMCID: PMC11937475 DOI: 10.1007/s00262-025-04003-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
Microwave ablation (MWA) is a super minimally invasive therapeutic approach that has been widely applied in the treatment of non-small cell lung cancer (NSCLC). Although MWA can elicit antitumor immune responses, these immune responses are not relatively steady and insufficient to completely clear recurrence tumor cells within the body. Immunotherapy monotherapy has shown low clinical efficacy in the treatment of advanced NSCLC. MWA combined with immune checkpoint inhibitors (ICIs) is a promising therapeutic approach. However, the mechanism of synergic effect remains elusive. In this study, we have conducted a retrospective analysis of the clinical outcomes of MWA combined with ICIs, finding that the combinational therapy yielded superior Objective Response Rate and longer Progression-Free Survival. In preclinical models, we established a tumor rechallenged model to address post-MWA recurrence and to delve into the underlying mechanisms of the combined therapy. We observed that the combined treatment (MWA + PD-L1 blockade therapy) effectively addressed the issue of tumor recurrence in tumor rechallenged model. The combinational therapy increased the function and percentage of CD8+ tumor-infiltrating lymphocytes, enhanced the functionality of CD8+ T cells within tumor-draining lymph nodes (TdLNs), and elevated the proportion of T central memory cells. Additionally, the combined treatments promoted the proportion of Migration Dendritic Cells type 1 (Mig DC1) within TdLNs, thereby enhancing their activation potential. Notably, FTY720-mediated blockade of lymphocyte egress abolished the therapeutic benefits, confirming TdLNs-dependent systemic immunity. Moreover, the efficacy of the combinational therapy depended on the migration of T cells from TdLNs to tumor site. In summary, we proposed a potentially effective combined treatment regimen and have elucidated the underlying cellular mechanisms that underpin its efficacy.
Collapse
Affiliation(s)
- Fengkuo Xu
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Jing Sang
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Nan Wang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Meixiang Wang
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | | | - Ji Ma
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Huanan Chen
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong. Engineering Laboratory for Health Management, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, 250014, China
- Department of Medical Record Management and Statistics, The First Affiliated Hospital of Shandong First Medical University &Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Qi Xie
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Shandong Provincial Lab for Clinical Immunology Translational Medicine in Universities, Jinan, 250014, China.
| | - Zhigang Wei
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| | - Xin Ye
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Shandong Provincial Lab for Clinical Immunology Translational Medicine in Universities, Jinan, 250014, China.
| |
Collapse
|
2
|
Su S, Wang Y, Lo EM, Tamukong P, Kim HL. High-intensity focused ultrasound ablation to increase tumor-specific lymphocytes in prostate cancer. Transl Oncol 2025; 53:102293. [PMID: 39862483 PMCID: PMC11803900 DOI: 10.1016/j.tranon.2025.102293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/27/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
Treatment options for localized prostate cancer have been expanded by FDA-approval of High-Intensity Focused Ultrasound (HIFU). Prostate cancer typically has few tumor-infiltrating lymphocytes, which are crucial for antitumor immunity. This study investigated the use of HIFU to increase lymphocyte infiltration into the tumor and enhance antitumor immunity. RM1 prostate tumors were implanted onto both flanks of syngeneic C57BL/6 J mice, with one tumor subjected to HIFU treatment. The growth of the contralateral tumor was monitored. Blood samples were obtained from patients both before and after prostatectomy or HIFU treatment. Peripheral blood mononuclear cells (PBMCs) were then isolated to analyze the immune cells. In murine experiments, the application of HIFU to one tumor decreased the growth of the contralateral (non-HIFU treated) tumor, when the contralateral tumor was the same tumor type, but not when it was a different tumor type. HIFU increased infiltration of CD4+ and CD8+ lymphocytes into the contralateral, same-type tumor. Lymphocyte depletion studies affirmed that the antitumor immune response triggered by HIFU relies on CD4+ and CD8+ lymphocytes. Addition of cholesterol-lowering intervention further increased antitumor immunity generated by HIFU in mice. In human subjects, HIFU, but not prostatectomy, stimulated anti-tumor CD4+ and CD8+ lymphocytes. We concluded that HIFU induced a potent cellular antitumor immune response that inhibited the progression of murine prostate tumors. HIFU stimulated tumor-specific cellular immunity in patients. Future clinical trials should explore the clinical benefits of HIFU, possibly in combination with existing immunotherapies, as immune modulators for both localized and metastatic disease.
Collapse
Affiliation(s)
- Shengchen Su
- Department of Urology, Cedars Sinai Medical Center, 8635 W. Third St, 1070, Los Angeles, CA 90048, United States
| | - Yanping Wang
- Department of Urology, Cedars Sinai Medical Center, 8635 W. Third St, 1070, Los Angeles, CA 90048, United States
| | - Eric M Lo
- Department of Urology, Cedars Sinai Medical Center, 8635 W. Third St, 1070, Los Angeles, CA 90048, United States
| | - Patrick Tamukong
- Department of Urology, Cedars Sinai Medical Center, 8635 W. Third St, 1070, Los Angeles, CA 90048, United States
| | - Hyung L Kim
- Department of Urology, Cedars Sinai Medical Center, 8635 W. Third St, 1070, Los Angeles, CA 90048, United States.
| |
Collapse
|
3
|
Bitar R, Salem R, Finn R, Greten TF, Goldberg SN, Chapiro J, Atzen S. Interventional Oncology Meets Immuno-oncology: Combination Therapies for Hepatocellular Carcinoma. Radiology 2024; 313:e232875. [PMID: 39560477 PMCID: PMC11605110 DOI: 10.1148/radiol.232875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 11/20/2024]
Abstract
The management of hepatocellular carcinoma (HCC) is undergoing transformational changes due to the emergence of various novel immunotherapies and their combination with image-guided locoregional therapies. In this setting, immunotherapy is expected to become one of the standards of care in both neoadjuvant and adjuvant settings across all disease stages of HCC. Currently, more than 50 ongoing prospective clinical trials are investigating various end points for the combination of immunotherapy with both percutaneous and catheter-directed therapies. This review will outline essential tumor microenvironment mechanisms responsible for disease evolution and therapy resistance, discuss the rationale for combining locoregional therapy with immunotherapy, summarize ongoing clinical trials, and report on developing imaging end points and novel biomarkers that are relevant to both diagnostic and interventional radiologists participating in the management of HCC.
Collapse
Affiliation(s)
- Ryan Bitar
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Riad Salem
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Richard Finn
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Tim F. Greten
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - S. Nahum Goldberg
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Julius Chapiro
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Sarah Atzen
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| |
Collapse
|
4
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
5
|
Shubhra QTH, Feczkó T, Cai Q. Beyond the danger signal: RNA aggregates orchestrate immunotherapy. Trends Mol Med 2024; 30:702-704. [PMID: 38866645 DOI: 10.1016/j.molmed.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
Mendez-Gomez et al. recently demonstrated the transformative potential of RNA-lipid particle aggregates (RNA-LPAs) in immunotherapy. By reprogramming the tumor microenvironment (TME) and potentiating antitumor immunity, RNA-LPAs target primary tumors and elicit robust systemic immunity. This innovative platform holds promise for translating preclinical success into tangible clinical benefits.
Collapse
Affiliation(s)
- Quazi T H Shubhra
- Institute of Chemistry, University of Silesia in Katowice, Szkolna 9, 40-003 Katowice, Poland; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Tivadar Feczkó
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; Faculty of Engineering, University of Pannonia, Egyetem u. 10, H-8200, Veszprém, Hungary
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
6
|
Cheng H, Lee W, Hsu F, Lai Y, Huang S, Lim CSH, Lin Z, Hsu S, Chiang C, Jeng L, Shyu W, Chen S. Manipulating the Crosstalk between Cancer and Immunosuppressive Cells with Phototherapeutic Gold-Nanohut for Reprogramming Tumor Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404347. [PMID: 38923327 PMCID: PMC11348132 DOI: 10.1002/advs.202404347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Photoimmunotherapy faces challenges due to insufficient intratumoral accumulation of photothermal agents and the reversion of the cancer-immunity cycle during treatment. In this study, an anti-PD-L1-immobilized magnetic gold nanohut, AuNH-2-Ab, with photoresponsive, thermosensitive, and immunomodulatory properties to effectively suppress the growth of primary tumors, elevate immunogenic cell death (ICD) levels, reverse the tumor immune microenvironment (TIME), and consequently inhibit metastases are developed. AuNH-2-Ab achieves high tumor accumulation (9.54% injected dose) following systemic administration, allowing the modulation of hyperthermia dose of over 50 °C in the tumor. By optimizing the hyperthermia dose, AuNH-2-Ab simultaneously target and eliminate cancer cells and tumor-associated macrophages, thereby activating potent antitumor immunity without being compromised by immunosuppressive elements. Hyperthermia/pH induced morphological transformation of AuNH-2-Ab involving the detachment of the surface antibody for in situ PD-L1 inhibition, and exposure of the inner fucoidan layer for natural killer (NK) cell activation. This precision photoimmunotherapy approach reprograms the TIME, significantly prolongs survival in a murine hepatocellular carcinoma model (Hep55.1c), and harnesses the synergistic effects of ICD production and checkpoint inhibitors by utilizing a single nanoplatform.
Collapse
Affiliation(s)
- Hung‐Wei Cheng
- Department of Materials Science and EngineeringNational Yang Ming Chiao Tung UniversityHsinchu30010Taiwan
| | - Wei Lee
- Cell Therapy CenterChina Medical University HospitalTaichung40447Taiwan
| | - Fei‐Ting Hsu
- Department of Biological Science and TechnologyChina Medical UniversityTaichung406040Taiwan
| | - Yen‐Ho Lai
- Cell Therapy CenterChina Medical University HospitalTaichung40447Taiwan
| | - Shu‐Rou Huang
- Translational Medicine Research CenterNew Drug development Center and Department of NeurologyChina Medical University HospitalTaichung40447Taiwan
| | - Chris Seh Hong Lim
- Department of Physician Assistant StudiesSchool of Health and Rehabilitation SciencesMGH InstituteBostonMassachusetts02114USA
| | - Zhen‐Kai Lin
- Department of Materials Science and EngineeringNational Yang Ming Chiao Tung UniversityHsinchu30010Taiwan
| | - Shih‐Chao Hsu
- Department of SurgeryChina Medical University HospitalTaichung40447Taiwan
| | - Chih‐Sheng Chiang
- Cell Therapy CenterChina Medical University HospitalTaichung40447Taiwan
- Graduate Institute of Biomedical ScienceChina Medical UniversityTaichung406040Taiwan
- Neuroscience and Brain Disease CenterChina Medical UniversityTaichung40447Taiwan
| | - Long‐Bin Jeng
- Cell Therapy CenterChina Medical University HospitalTaichung40447Taiwan
- Organ Transplantation CenterChina Medical University HospitalTaichung40447Taiwan
- School of MedicineChina Medical UniversityTaichung406040Taiwan
| | - Woei‐Cherng Shyu
- Translational Medicine Research CenterNew Drug development Center and Department of NeurologyChina Medical University HospitalTaichung40447Taiwan
- Graduate Institute of Biomedical ScienceChina Medical UniversityTaichung406040Taiwan
- Neuroscience and Brain Disease CenterChina Medical UniversityTaichung40447Taiwan
| | - San‐Yuan Chen
- Department of Materials Science and EngineeringNational Yang Ming Chiao Tung UniversityHsinchu30010Taiwan
- Graduate Institute of Biomedical ScienceChina Medical UniversityTaichung406040Taiwan
- School of DentistryCollege of Dental MedicineKaohsiung Medical UniversityKaohsiung807Taiwan
| |
Collapse
|
7
|
Mao J, Liu L, Shen Q, Cen M. Integrating single-cell transcriptomics and machine learning to predict breast cancer prognosis: A study based on natural killer cell-related genes. J Cell Mol Med 2024; 28:e18549. [PMID: 39098994 PMCID: PMC11298315 DOI: 10.1111/jcmm.18549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in women globally. Natural killer (NK) cells play a vital role in tumour immunosurveillance. This study aimed to establish a prognostic model using NK cell-related genes (NKRGs) by integrating single-cell transcriptomic data with machine learning. We identified 44 significantly expressed NKRGs involved in cytokine and T cell-related functions. Using 101 machine learning algorithms, the Lasso + RSF model showed the highest predictive accuracy with nine key NKRGs. We explored cell-to-cell communication using CellChat, assessed immune-related pathways and tumour microenvironment with gene set variation analysis and ssGSEA, and observed immune components by HE staining. Additionally, drug activity predictions identified potential therapies, and gene expression validation through immunohistochemistry and RNA-seq confirmed the clinical applicability of NKRGs. The nomogram showed high concordance between predicted and actual survival, linking higher tumour purity and risk scores to a reduced immune score. This NKRG-based model offers a novel approach for risk assessment and personalized treatment in BC, enhancing the potential of precision medicine.
Collapse
Affiliation(s)
- Juanjuan Mao
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Ling‐lin Liu
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Qian Shen
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| | - Mengyan Cen
- Department of Thyroid and Breast SurgeryNingbo Hospital of TCM Affiliated to Zhejiang Chinese Medicine UniversityNingbo CityZhejiang ProvinceChina
| |
Collapse
|
8
|
Li N, Geng S, Dong ZZ, Jin Y, Ying H, Li HW, Shi L. A new era of cancer immunotherapy: combining revolutionary technologies for enhanced CAR-M therapy. Mol Cancer 2024; 23:117. [PMID: 38824567 PMCID: PMC11143597 DOI: 10.1186/s12943-024-02032-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024] Open
Abstract
Significant advancements have been made in the application of chimeric antigen receptor (CAR)-T treatment for blood cancers during the previous ten years. However, its effectiveness in treating solid tumors is still lacking, necessitating the exploration of alternative immunotherapies that can overcome the significant challenges faced by current CAR-T cells. CAR-based immunotherapy against solid tumors shows promise with the emergence of macrophages, which possess robust phagocytic abilities, antigen-presenting functions, and the ability to modify the tumor microenvironment and stimulate adaptive responses. This paper presents a thorough examination of the latest progress in CAR-M therapy, covering both basic scientific studies and clinical trials. This study examines the primary obstacles hindering the realization of the complete potential of CAR-M therapy, as well as the potential strategies that can be employed to overcome these hurdles. With the emergence of revolutionary technologies like in situ genetic modification, synthetic biology techniques, and biomaterial-supported gene transfer, which provide a wider array of resources for manipulating tumor-associated macrophages, we suggest that combining these advanced methods will result in the creation of a new era of CAR-M therapy that demonstrates improved efficacy, safety, and availability.
Collapse
Affiliation(s)
- Na Li
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
- Department of Immunology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Shinan Geng
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
| | - Zhen-Zhen Dong
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ying Jin
- Hangzhou Institute of Medicine (HIM), Zhejiang Caner Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hangjie Ying
- Hangzhou Institute of Medicine (HIM), Zhejiang Caner Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hung-Wing Li
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Liyun Shi
- Key lab of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China.
| |
Collapse
|
9
|
Jeon HJ, Chun HJ, Choi HS, Keum B, Kim HB, Kim JH. Biphasic Regulation of Apoptosis Following Gastric Irreversible Electroporation Using Tissue Immunohistochemistry of Activated Caspase-3 with TUNEL Method. Cancers (Basel) 2024; 16:1389. [PMID: 38611067 PMCID: PMC11010973 DOI: 10.3390/cancers16071389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
The regulation of apoptosis is the primary goal of ablation therapy. Irreversible electroporation (IRE) is a promising non-thermal tissue ablation-based therapy that induces apoptosis by manipulating electrical conditions. This study aimed to investigate IRE-induced gastric tissue apoptosis in response to changes in the electric field intensity, followed by the repair process. Among the 52 rats used in this study, 24 were used to explore apoptosis, and 28 were used to study regeneration. The apoptosis-to-necrosis ratio of the electrical field strength was evaluated using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling and caspase-3 immunohistochemistry. The size of IRE-induced ulcers in the gastric tissue continuously increased with increasing electrical intensity (r2 = 0.830, p < 0.001). The level of apoptosis gradually decreased after peaking at 200 V (1000 V/cm). The size of the 400 V-ablated ulcers continued to decrease, and they were not visible by day 14. The proliferation and migration of epithelial cells with fibroblasts were observed on day 3 and augmented on day 7 post-ablation. This investigation demonstrated the biphasic activation of apoptosis with respect to the electrical field strength. Visually and histologically, IRE-induced gastric ulcers demonstrated complete tissue regeneration after two weeks.
Collapse
Affiliation(s)
- Han Jo Jeon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.); (B.K.)
| | - Hoon Jai Chun
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.); (B.K.)
| | - Hyuk Soon Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.); (B.K.)
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.); (B.K.)
| | - Hong Bae Kim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea;
| | - Jong Hyuk Kim
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
| |
Collapse
|
10
|
Deng D, Hao T, Lu L, Yang M, Zeng Z, Lovell JF, Liu Y, Jin H. Applications of Intravital Imaging in Cancer Immunotherapy. Bioengineering (Basel) 2024; 11:264. [PMID: 38534538 DOI: 10.3390/bioengineering11030264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/20/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Currently, immunotherapy is one of the most effective treatment strategies for cancer. However, the efficacy of any specific anti-tumor immunotherapy can vary based on the dynamic characteristics of immune cells, such as their rate of migration and cell-to-cell interactions. Therefore, understanding the dynamics among cells involved in the immune response can inform the optimization and improvement of existing immunotherapy strategies. In vivo imaging technologies use optical microscopy techniques to visualize the movement and behavior of cells in vivo, including cells involved in the immune response, thereby showing great potential for application in the field of cancer immunotherapy. In this review, we briefly introduce the technical aspects required for in vivo imaging, such as fluorescent protein labeling, the construction of transgenic mice, and various window chamber models. Then, we discuss the elucidation of new phenomena and mechanisms relating to tumor immunotherapy that has been made possible by the application of in vivo imaging technology. Specifically, in vivo imaging has supported the characterization of the movement of T cells during immune checkpoint inhibitor therapy and the kinetic analysis of dendritic cell migration in tumor vaccine therapy. Finally, we provide a perspective on the challenges and future research directions for the use of in vivo imaging technology in cancer immunotherapy.
Collapse
Affiliation(s)
- Deqiang Deng
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tianli Hao
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lisen Lu
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Muyang Yang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen Zeng
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Yushuai Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
11
|
Pastori C, Nafie EHO, Wagh MS, Mammarappallil JG, Neal RE. Pulsed Electric Field Ablation versus Radiofrequency Thermal Ablation in Murine Breast Cancer Models: Anticancer Immune Stimulation, Tumor Response, and Abscopal Effects. J Vasc Interv Radiol 2024; 35:442-451.e7. [PMID: 38042523 DOI: 10.1016/j.jvir.2023.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023] Open
Abstract
PURPOSE To compare the immune response and survival after size-matched radiofrequency (RF) ablation and a proprietary form of pulsed electric field (PEF) ablation in murine tumors. MATERIAL AND METHODS Orthotopically inoculated EMT6 or 4T1 murine tumors received sham, RF ablation, or PEF ablation. 4T1 tumor ablations included subgroups with intraperitoneal checkpoint inhibition immunotherapy (αPD-1). Blood was collected for cytokine profiling and flow cytometry. Tumor size was measured and survival was monitored. Tumor samples were processed for histology, immunohistochemistry, flow cytometry, and cytokine profiling. Lungs were collected from 4T1-bearing mice for hematoxylin and eosin histology to assess metastatic spread and abscopal effect induced by ablation. RESULTS PEF elicited distinct immunomodulatory effects, with clear differences in serum and tumor cytokine profiles compared with RF ablation, including intratumoral downregulation of vascular endothelial growth factor, hypoxia-inducible factor 1α, c-MET, interleukin-10, Ki67, and tumor necrosis factor-α (all P < .05). PEF increased innate immune activation, with enhanced recruitment of dendritic cells, M1 macrophages, and natural killer cells coupled with a reduction in M2 macrophages and myeloid-derived suppressor cells (all P < .05). Concurrently, PEF strengthened adaptive immunity compared with RF ablation, characterized by increased antigen-specific T cells and decreased regulatory T cells (all P < .05). PEF stalled tumor growth and increased survival at the end of the study (≥4× versus RFA). Finally, PEF promoted an abscopal effect of clearing metastases in the lungs, which was stronger in combination with αPD-1 than with PEF alone. CONCLUSIONS The proprietary form of PEF used in this study evoked a preferential immunostimulatory profile versus RF ablation thermal ablation in mice, with implications for enhancing the therapeutic effectiveness of checkpoint inhibition immunotherapy for immunotherapy-unresponsive tumors.
Collapse
Affiliation(s)
- Chiara Pastori
- Department of Research, Galvanize Therapeutics, Redwood City, California
| | - Ebtesam H O Nafie
- Department of Research, Galvanize Therapeutics, Redwood City, California
| | - Mukta S Wagh
- Department of Research, Galvanize Therapeutics, Redwood City, California
| | | | - Robert E Neal
- Department of Research, Galvanize Therapeutics, Redwood City, California.
| |
Collapse
|
12
|
Qian L, Xie L, Zhu Y, Huang C, Meng Z. Potent induction of antitumor immunity by combining cryo-thermal ablation with immune checkpoint inhibitors in hepatocellular carcinoma. Liver Int 2024; 44:723-737. [PMID: 38111027 DOI: 10.1111/liv.15817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 11/08/2023] [Accepted: 12/02/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND The low response rate of immune checkpoint inhibitors (ICIs) prompts the exploration of novel combination therapies for patients with hepatocellular carcinoma (HCC). Here, we aimed to examine the efficiency and potential mechanism of cryo-thermal ablation (Cryo-A) combined with anti-programmed death protein 1 (αPD1) and/or cytotoxic T-lymphocyte antigen 4 (αCTLA4) inhibitors in a murine hepatoma model. METHOD Immunocompetent C57BL/6 mice inoculated with unilateral or bilateral H22 hepatic tumour cells were treated with Cryo-A and/or ICIs (αPD1 and/or αCTLA4). Flow cytometry, immunohistochemistry, ELISpot assay, time-of-flight cytometry, tumour rechallenging, and T-cell depletion assay were used to assess the dynamic changes of immune cell subsets following therapy. RESULTS We found Cryo-A resulted in immunogenic cell death of tumour cells, activation of dendritic cells, and enhancement of antitumor immunity. Cryo-A alone was insufficient to extend survival, combining Cryo-A with αPD1 and αCTLA4 further modulated the tumour microenvironment, inducing a durable antitumor immune response by tumour-reactive CD8+ T cells and significantly prolonged survival. Time-of-flight cytometry (CyTOF) data revealed that combination therapies reshaped the tumour microenvironment by the increase of intratumoral CD8+ T cells expressed higher levels of cytotoxic markers and immune checkpoint molecules, and by downregulation of intratumoral granulocytes. The combination also resulted in the eradication of remote unablated tumours (abscopal effect). CONCLUSIONS These findings suggested that Cryo-A turned HCC from "cold" tumours to "hot" tumours and the combination of Cryo-A with αPD1 and αCTLA4 may be a promising approach to improve the prognosis of HCC.
Collapse
Affiliation(s)
- Ling Qian
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Zhu
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changjing Huang
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Jin S, Guo Y, Wang X. Development of Platinum Complexes for Tumor Chemoimmunotherapy. Chemistry 2024; 30:e202302948. [PMID: 38171804 DOI: 10.1002/chem.202302948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Indexed: 01/05/2024]
Abstract
Platinum complexes are potential antitumor drugs in chemotherapy. Their impact on tumor treatment could be greatly strengthened by combining with immunotherapy. Increasing evidences indicate that the antitumor activity of platinum complexes is not limited to chemical killing effects, but also extends to immunomodulatory actions. This review introduced the general concept of chemoimmunotherapy and summarized the progress of platinum complexes as chemoimmunotherapeutic agents in recent years. Platinum complexes could be developed into inducers of immunogenic cell death, blockers of immune checkpoint, regulators of immune signaling pathway, and modulators of tumor immune microenvironment, etc. The synergy between chemotherapeutic and immunomodulatory effects reinforces the antitumor activity of platinum complexes, and helps them circumvent the drug resistance and systemic toxicity. The exploration of platinum complexes for chemoimmunotherapy may create new opportunities to revive the discovery of metal anticancer drugs.
Collapse
Affiliation(s)
- Suxing Jin
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Yan Guo
- School of Materials and Chemical Engineering, Henan University of Urban Construction, Pingdingshan, 467036, Henan, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
14
|
Ruff SM, Pawlik TM. Editorial on immune checkpoint inhibitors in the treatment of hepatocellular carcinoma. Immunotherapy 2023; 15:1323-1326. [PMID: 37694380 DOI: 10.2217/imt-2023-0161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Affiliation(s)
- Samantha M Ruff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center & James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center & James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Sun S, Zhang Y, Li Y, Wei L. Crosstalk between colorectal cancer cells and cancer-associated fibroblasts in the tumor microenvironment mediated by exosomal noncoding RNAs. Front Immunol 2023; 14:1161628. [PMID: 37234178 PMCID: PMC10206140 DOI: 10.3389/fimmu.2023.1161628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Colorectal cancer (CRC) is a common malignant tumor of the digestive system, and its morbidity rates are increasing worldwide. Cancer-associated fibroblasts (CAFs), as part of the tumor microenvironment (TME), are not only closely linked to normal fibroblasts, but also can secrete a variety of substances (including exosomes) to participate in the regulation of the TME. Exosomes can play a key role in intercellular communication by delivering intracellular signaling substances (e.g., proteins, nucleic acids, non-coding RNAs), and an increasing number of studies have shown that non-coding RNAs of exosomal origin from CAFs are not only closely associated with the formation of the CRC microenvironment, but also increase the ability of CRC to grow in metastasis, mediate tumor immunosuppression, and are involved in the mechanism of drug resistance in CRC patients receiving. It is also involved in the mechanism of drug resistance after radiotherapy in CRC patients. In this paper, we review the current status and progress of research on CAFs-derived exosomal non-coding RNAs in CRC.
Collapse
Affiliation(s)
| | | | | | - Linlin Wei
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
16
|
PCSK9 Inhibitors in Cancer Patients Treated with Immune-Checkpoint Inhibitors to Reduce Cardiovascular Events: New Frontiers in Cardioncology. Cancers (Basel) 2023; 15:cancers15051397. [PMID: 36900189 PMCID: PMC10000232 DOI: 10.3390/cancers15051397] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cancer patients treated with immune checkpoint inhibitors (ICIs) are exposed to a high risk of atherosclerosis and cardiometabolic diseases due to systemic inflammatory conditions and immune-related atheroma destabilization. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key protein involved in metabolism of low-density lipoprotein (LDL) cholesterol. PCSK9 blocking agents are clinically available and involve monoclonal antibodies, and SiRNA reduces LDL levels in high-risk patients and atherosclerotic cardiovascular disease events in multiple patient cohorts. Moreover, PCSK9 induces peripheral immune tolerance (inhibition of cancer cell- immune recognition), reduces cardiac mitochondrial metabolism, and enhances cancer cell survival. The present review summarizes the potential benefits of PCSK9 inhibition through selective blocking antibodies and siRNA in patients with cancer, especially in those treated with ICIs therapies, in order to reduce atherosclerotic cardiovascular events and potentially improve ICIs-related anticancer functions.
Collapse
|